Targeted Nanoparticles ~drug delivery~kanai/seminar/pdf/Lit_S_Ito_M1.pdf · Targeted Polymeric...

Post on 16-Mar-2020

10 views 0 download

Transcript of Targeted Nanoparticles ~drug delivery~kanai/seminar/pdf/Lit_S_Ito_M1.pdf · Targeted Polymeric...

Targeted Polymeric Nanoparticles

~drug delivery~

Literature Seminar

May 21, 2012

Soichi Ito (M1)

1

Contents

I. Introduction

II. Topics

1. Passive vs Active targeting

2. Preparation of targeted polymeric NPs

3. Targeting ligands

4. Optimal biophysicochemical characteristics

III.Perspective

2

Historical timeline of clinical-stage

nanoparticle technologies

3 JINJUN SHI et al. ACCOUNTS OF CHEMICAL RESEARCH, 2011, 44(10), 1123–1134.

• Polymeric NPs have the capability to

1. release drugs at an experimentally predetermined

rate over a prolonged period of time,

2. release drugs preferentially at target sites with the

possibility of controlled release rates,

3. maintain drug concentrations within therapeutically

appropriate ranges in circulation and within tissues,

4. protect drugs from hepatic inactivation, enzymatic

degradation and rapid clearance in vivo.

4

Targeted polymeric NPs

5 JINJUN SHI et al. ACCOUNTS OF CHEMICAL RESEARCH, 2011, 44(10), 1123–1134.

Targeted NPs in clinical development

6 Nazila Kamaly et al. Chem. Soc. Rev., 2012, 41, 2971–3010.

What is the targeted polymeric NPs?

poly(lactic-co-glycolic acid) (PLGA), poly(ethylene glycol) (PEG)

7 Nazila Kamaly et al. Chem. Soc. Rev., 2012, 41, 2971–3010.

Biodegradable polymers

poly(lactic acid) (PLA), poly(glycolic acid) (PGA),

poly(lactic-co-glycolic acid) (PLGA), poly(caprolactone) (PCL)

“Controlled Drug Release”

8 Nazila Kamaly et al. Chem. Soc. Rev., 2012, 41, 2971–3010.

Drug release mechanisms

a. Diffusion from polymer

matrix

b. Surface erosion/degradation

of polymer matrix

c. Biodegradation of polymer

matrix due to hydrolytic

degradation

9 Nazila Kamaly et al. Chem. Soc. Rev., 2012, 41, 2971–3010.

“Stealth” Nanopartile

The non-specific binding of plasma proteins onto

the surface of NPs, also known as opsonization,

leads to enhanced blood clearance by the cells

of mononulear phagocytic system (MPS).

By decorating the surfaces of NPs with PEG

polymers, the circulation times can be prolonged.

10

Contents

I. Introduction

II. Topics

1. Passive vs Active targeting

2. Preparation of targeted polymeric NPs

3. Targeting ligands

4. Optimal biophysicochemical characteristics

III.Perspective

11

EPR effect (Passive targeting) “Enhanced Permeation and Retention” effect

12 Pavan P. Adiseshaiah et al. WIREs Nanomed Nanobiotechnol 2009, 2, 99–112.

Limitations of Passive targeting

• Passively targeted NPs end up releasing their

therapeutic payload into the tumor milieu rather

than within cancer cells. (“PEG dilemma”)

• For drugs that are not readily retained in tumors or

macromolecular drugs that are not readily taken up

by cancer cells, this extracellular drug release may

be less effective at maintaining a differentially high

tumor drug concentration over an extended period

of time.

13

Passive vs active targeting

14 Omid C. Farokhzad et al. ACS Nano, 2009, 3 (1), 16–20.

Active targeting

• Targeted NPs facilitate receptor-mediated

endocytosis(RME), releasing therapeutic agents

inside target cell.

Higher therapeutic efficacy

Lower toxicity

15

Contents

I. Introduction

II. Topics

1. Passive vs Active targeting

2. Preparation of targeted polymeric NPs

3. Targeting ligands

4. Optimal biophysicochemical characteristics

III.Perspective

16

NP Formulation Method

“Bottom-up” (self-assembly)

Bulk synthesis

• Nanoprecipitation

• Oil-in-water emulsification-solvent evaporation

• Water-in-oil-in-water emulsification-solvent evaporation, etc.

Microfluidic synthesis

“Top-down”

PRINT

(Particle Replication In Non-wetting Templates) 17

Nanoprecipitation

Difficulty in complete removal of the organic solvent after self-

assembly.

18 Pegi Ahlin Grabnar et al. Journal of Microencapsulation 2011, 28(4), 323–335.

Single emulsion

• This method results in higher drug loading and

encapsulation efficiency compared to nanoprecipitation,

as well as achieving complete solvent removal.

• Obtained NPs are often larger than those obtained

through nanoprecipitation.

19

Double emulsion

• This method is generally used for encapsulation of hydrophilic drugs

• This method normally yields NPs with larger size than

nanoprecipitaion or O/W methods, with moderate drug loading and

encapsulation efficiency.

20

Guilin Wang et al. Expert Opin. Drug Deliv. 2008, 5(5), 499-515.

Microfluidic methods

21 Rohit Karnik et al. Nano Lett. 2008, 8(9), 2906-2912.

22 MARY E. NAPIER et al. Polymer Reviews, 2007, 47, 321–327.

23 Jin Wang et al. small, 2011, 7, No. 14, 1919–1931.

Drug loading methods

Encapsulation method is the most common technique in this field.

*The drug is entrapped in the polymer matrix during preparation of NPs.

24

Incorporation of targeting ligands on NPs

Coupling chemistry should

not lead to undesirable products or side reactions

be produced on large-scales in a reproducible manner 25

Post-synthesis NP surface modification method • Amide bond formation

• Maleimide coupling with thiols

• “Bioorthogonal” reactions such as

Cu-free click reactions

[4+2] cycloadditon reaction

26 Mariagrazia Di Marco et al. International Journal of Nanomedicine, 2010, 5, 37–49.

John C. Jewett et al. Chem. Soc. Rev., 2010, 39, 1272–1279.

W. Russ Algar et al. Bioconjugate Chem. 2011, 22, 825–858.

• It is difficult to control the stoichiometry of functional biomolecules

on the surface of NPs via coupling chemistries.

• Self-assembly of pre-functionalized triblock copolymers allows

for the reproducible creation of optimal targeted NPs.

Targeted NPs through polymer self-assembly

27 Frank Gu et al. PNAS, 2008, 105, 2586-2591.

Precisely controlled aptamer density

28

The effect of Apt surface density on NP

in vivo vs in vitro

29

Solubility of ligands

30 Pedro M. Valencia et al. Biomaterials, 2011, 32, 6226-6233.

Contents

I. Introduction

II. Topics

1. Passive vs Active targeting

2. Preparation of targeted polymeric NPs

3. Targeting ligands

4. Optimal biophysicochemical characteristics

III.Perspective

31

Targeting ligands

Antibodies and their fragments

Proteins

Peptides

Aptamers (Nucleic acid lidands)

Small molecules (folic acid, carbohydrate etc.)

32

Antibodies and their fragments

Type MW/kDa Diameter/nm

Whole antibodies 150 15-20

Fab’ 50 5-10

ScFv 25 3-5

Nanobody 15 2-3 33

Dan Peer et al. nature nanotechnology, 2007, 2, 751-760.

Proteins • Endogenous proteins that selectively bind to specific

membrane-bound receptors on cells can be used.

Transferrin, Epidermal Growth Factor, Nerve Growth Factor, etc.

The receptors of Tf and EGF are overexpressed on cancer cells.

Demerits

Commonly immunogenic, off-target adverse effects

34

Ulrich E. Schaible et al. NATURE REVIEWS, 2004, 2, 946-953.

Peptides

• Small size, relatively low immunogenicity, high

stability, and ease of conjugation to NP surfaces

RGD (Arg-Gly-Asp) sequence binds to αvβ3

integrin receptors which are highly upregurated on

both tumor cells and angiogenic endothelial cells.

Cell-penetrating peptides such as Tat peptide

Tat peptide derives from the HIV-1 virus.

Peptides with R/KXXR/K motif such as iRGD

iRGD homes to tumors and penetrates into them.

35

36 Kazuki N. Sugahara et al. Cancer Cell, 2009, 16, 510–520.

Aptamers

• Single-strand of DNA or RNA oligonucleotides

• Small size, reproducible synthesis, low immunity

Type MW/kDa Diameter/nm

Whole antibodies 150 15-20

Nanobody 15 2-3

Aptamers DNA/RNA 10-30 2-3

The high specificity of Apts against targets is their

secondary structure, but the secondary structure

may be affected by heat, exonuclease or

endonuclease degradation. 38

“cell-uptake selection”

39

Z. Xiao et al. ACS Nano, 2012, 6, 696–704.

Small molecules

• The availability of a range of facile coupling

chemistries for their conjugation

• The availability of a wide range of targeting ligands

with variable solubilities and functional groups

Folic acid (or folate)

Folate receptors (FRs) are frequently over-expressed

in a range of cancer

FRs are expressed not only in tumor tissue but in

normal epithelia.

40

Contents

I. Introduction

II. Topics

1. Passive vs Active targeting

2. Preparation of targeted polymeric NPs

3. Targeting ligands

4. Optimal biophysicochemical characteristics

III.Perspective

41

Influence of particle size

• The generally accepted diameter of nanomedicine

for cancer is in the range of 10-100 nm.

The lower limit is determined by an interaction

with renal filtration in the kidney.

The upper limit is determined by an interaction

with RES (immune system) in the spleen and liver. (particles larger than 200 nm must compensate by deformability)

For the purpose of tumor accumulation, the upper

limit for extravasation into solid tumors have been

suggested at ~400 nm.

42

• Influence of NP shape

Spheres vs Rods on cellular uptake?

Further investigations are required.

• Influence of NP surface charge

NP surface charge is a major factor contributing

to the non-specific binding of NPs to cells.

Charged NPs will inevitably have short half-lives

and high non-specific cellular uptakes due to

interaction with blood proteins and complement

activation.

Neutral particles would be good. 43

Influence of NP PEGylation

“mushroom”

“brush”

Optimal PEG coverage?

44 Donald E. Owens III et al. International Journal of Pharmaceutics, 2006, 307, 93–102.

Contents

I. Introduction

II. Topics

1. Passive vs Active targeting

2. Preparation of targeted polymeric NPs

3. Targeting ligands

4. Optimal biophysicochemical characteristics

III.Perspective

45

• Delivering therapeutics in a more controlled and

specific manner

Improved drug safety and efficacy

• Protecting drugs from rapid metabolism and

inactivation; improving drug solubility, PK, BD,

and target tissue exposure

Additional degrees of freedom to medical chemistry

“A new class of therapeutics”

46

Co-delivering of multiple drugs

47 N. Kolishetti et al. Proc. Natl. Acad. Sci. U. S. A., 2010, 107, 17939–17944.

Challenges Insufficient understanding of events at the nano-bio

interface in vitro and in vivo

Inadequate knowledge of the fate of NPs at the body, organ, and cellular levels

Difficulty in achieving reproducible and controlled synthesis of NPs at scales suitable for clinical development and commercialization

Overreliance on the EPR effect (This phenomenon may not be a universal property of all tumors.)

There are too many “on a case-by-case basis”.

Is it possible for a reasonable strategy to exist?

48

Reference • Nazila Kamaly et al. Chem. Soc. Rev., 2012, 41, 2971–3010. • JINJUN SHI et al. ACCOUNTS OF CHEMICAL RESEARCH, 2011, 44(10), 1123–1134. • Pavan P. Adiseshaiah et al. WIREs Nanomed Nanobiotechnol 2009, 2, 99–112. • Omid C. Farokhzad et al. ACS Nano, 2009, 3 (1), 16–20. • Pegi Ahlin Grabnar et al. Journal of Microencapsulation 2011, 28(4), 323–335. • Guilin Wang et al. Expert Opin. Drug Deliv. 2008, 5(5), 499-515. • Rohit Karnik et al. Nano Lett. 2008, 8(9), 2906-2912. • MARY E. NAPIER et al. Polymer Reviews, 2007, 47, 321–327. • Jin Wang et al. small, 2011, 7, No. 14, 1919–1931. • Mariagrazia Di Marco et al. International Journal of Nanomedicine, 2010, 5, 37–49. • John C. Jewett et al. Chem. Soc. Rev., 2010, 39, 1272–1279. • Frank Gu et al. PNAS, 2008, 105, 2586-2591. • Pedro M. Valencia et al. Biomaterials, 2011, 32, 6226-6233. • Dan Peer et al. nature nanotechnology, 2007, 2, 751-760. • Ulrich E. Schaible et al. Nature Reviews Microbiology 2004, 2, 946-953. • Kazuki N. Sugahara et al. Cancer Cell 2009, 16, 510–520. • W. Russ Algar et al. Bioconjugate Chem. 2011, 22, 825–858. • Z. Xiao et al. ACS Nano, 2012, 6, 696–704. • Donald E. Owens III et al. International Journal of Pharmaceutics, 2006, 307, 93–102. • N. Kolishetti et al. Proc. Natl. Acad. Sci. U. S. A., 2010, 107, 17939–17944.

• http://www.creative-biolabs.com/phagedisplay1.htm • wikipedia

49